Abstract

Single-cell RNA sequencing (scRNA-seq) technology has made great strides in research over the last decade. Data analysis has been aided by developments in bioinformatics tools and artificial intelligence, allowing biological and clinical researchers to get a deeper understanding of the different cell clusters and their dynamics within tumours. Combining conventional treatment modalities like chemotherapy and radiation with immunotherapy is a growing trend in cancer treatment. Hence, knowledge of the tumour microenvironment and the effect of each treatment modality on the TME, at a single cell level can provide treating clinicians with better clues for patient stratification and prognostication. With this knowledge, immunotherapy could become successful in treating a wide range of cancers, opening the path for the creation of even more effective treatment strategies. Despite the widespread availability of scRNA-seq technology, computational analysis and data interpretation are still challenges. Worldwide, such challenges are being addressed by various researchers, strengthening the contribution of this technology towards cancer elimination. In this mini-review, we primarily focus on the technique, its workflow, and the computational aspects of scRNA technology, along with an overview of the current challenges in the analysis and interpretation of the data generated.
Introduction

An important role in tumour development and heterogeneity is played by the tumour microenvironment (TME), which is the environment around the tumour consisting of the extracellular matrix, fibroblasts, and immune cells, along with the blood vessels surrounding it [1, 2]. Through the mechanism of cellular communication, all the cell types within the tumour microenvironment interact with each other to promote tumour development [3]. Hence, the implementation of effective anticancer immunotherapeutic strategies requires an understanding of the crucial suppressive mechanisms of the TME [4, 5]. To assess and track the changes within the tumour, next-generation sequencing (NGS) methods can be used. NGS is a high-throughput sequencing technology used to analyse DNA and RNA samples at high speed and low cost. Single-cell RNA sequencing (scRNA-seq), an offshoot of the NGS technology, can provide information on a combination of genomic, transcriptomic, epigenomic, and immunological signatures at the level of a single cell [6]. In this mini-review, we aim to provide a brief overview of the pros and cons of scRNA technology and its use in cancer immunotherapy.
Overview of scRNA sequencing steps
Rare cell mutations that occur during the processes of tumour progression, invasion, metastasis, and activation of immune cells can be monitored by using the scRNA sequencing method [7, 8]. In order to detect the transcriptional activity of several genes, including mutated ones and those that act as immune checkpoints, scRNA-Seq can be combined with traditional techniques like flow cytometry to specifically screen for targets of immune modulation. In brief, the initial step in scRNA-seq is the isolation of single cells from a heterogeneous cell population, followed by the extraction of ribonucleic acid (RNA) from the single cell, barcoded reverse transcription, cDNA amplification, library preparation, and sequencing the library using NGS platforms like the C1 single cell autoprep system (Fluidigm) or chromium (10× genomics) [9], [10], [11], [12]. A comprehensive workflow for scRNA sequencing and analysis is provided in Figure 1.

An overview of the SCRNA sequencing workflow and its application in immunotherapy. It was created by using BioRender.com.
Various techniques and platforms used in ScRNA sequencing
Isolation of the cells can be done by various methods, depending on the type of sample and its processing. The techniques used for single-cell isolation require skill and knowledge of cell morphology. There are two ways of cell isolation based on their physical properties and their cellular biological characteristics [13]. Here we have summarised various techniques used to isolate the single cell and their application in Table 1.
Various methods of single-cell preparation.
Method of single cell preparation | Application | Ref. |
---|---|---|
Fluorescence-activated cell sorting (FACS) | This technique enables the concurrent assessment of several parameters, both quantitative and qualitative, in individual cells. The process of FACS sorting may effectively segregate molecules based on their varying levels of expression. | [14, 15] |
Magnetically activated cell sorting (MACS) | The MACS technology exhibits a relative simplicity and cost-effectiveness. The functionality of MACS devices relies on the specificity and affinity of the antibodies used for the purpose of isolating the desired target cells. One limitation of this approach is its reliance on cell surface chemicals as the only indicators for the isolation of viable cells. The immunomagnetic methods used in this method restrict its capabilities compared to FACS, since they alone enable the separation of cells into positive and negative populations. | [14, 16] |
Laser-capture microdissection (LCM) | To optimise the use of developing molecular analytical methods like as PCR, microarrays, and proteomics, it is essential to effectively and precisely identify and capture the specific cells of interest. One significant constraint is the need to visually examine cells of interest using microscopic examination of morphological traits. This requires the expertise of a qualified professional such as a pathologist, cytologist, or technologist who is skilled in cell identification. | [17, 18] |
Manual cell picking and micromanipulation | The execution of micromanipulation techniques may be readily accomplished inside an electrophysiology laboratory that is equipped with a patch clamp device. Nevertheless, the throughput is constrained, necessitating the involvement of proficient experts for its execution. The detection of complicated changes is subject to limitations in terms of its usefulness. | [14, 19] |
Microfluidics | It is possible to integrate many separation techniques, including filtration and sedimentation, as well as affinity-based methodologies such as fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS). Microfluidic chips have promising prospects in the fields of DNA sequencing, protein analysis, cell manipulation, and cell composition analysis. | [20] |
Cell celector platform | Platforms are used for the purpose of segregating circulating cancer cells, and individual foetal cells for cell-based non-invasive prenatal diagnostics, antibody exploration, and heterogeneity analysis. | [14, 21] |
In the 10× Genomics platform, single cell suspensions are loaded onto gel beads containing the ingredients for reverse transcription along with cell and transcript-identifying barcodes [22]. This combination is unique as it can be used to distinguish the expression profile of each cell. Based on this combination, the depth of the sequencing step is calculated [13]. Approximately 20,000 cells with a multiplexing possibility of 12 samples per lane [23]. With this input, 4.8 billion reads can be achieved in depth. Either the Miseq or Hiseq platforms can be used to perform the scRNA sequencing [24]. Both gene expression (GEX) and T cell/B cell repertoire profiling can be combined for the same input of cells using the 10× platform.
Computational tools and pipelines for scRNA sequencing analysis
Choosing the right tool for scRNA sequencing data analysis is a major task. The output file generated by scRNA sequencing through the Illumina HiSeq/MiSeq platform is the Base Call (BCL) file. The first step in data analysis will be BCL to FastQ conversion using tools like “bcl2fastQ”, and CASAVA. The quality control for the FastQ file is evaluated by an open-source command-line tool called “FASTQC”. After FASTQC, the output files can be analysed for an application of our interest using different tools [25]. Quantifying gene expression, identifying cell populations, immune repertoire studies, single-cell resolution and spatial location are the areas of interest in the ScRNA data analysis. Spatial data can be analyzed by using Squidpy, Seurat, Giotto, and spatial experiment tools to view the resolution spatially [26]. Tools like Scirpy, Dandelion, and scRepertoire can help with adaptive immune receptor repertoire analysis using the ScRNA-seq dissociated data. Muon, Seurat, and CiteFuse are used to know the functions of surface proteins. Muon, ArchR, snapATAC, Signac, Scanpy, Seurat, and single-cell experiments are used to measure chromatin accessibility and intracellular transcriptome gene quantity [25, 27]. In parallel, 10× genomics developed an ScRNA analysis pipeline called the Cell Ranger, which provides comprehensive solutions for processing and analysing data generated using the 10× Genomics chromium platform [25, 28].
Applications of scRNA sequencing in immunotherapy
Effective immunotherapeutic strategies require suitable target antigens that facilitate optimal stimulation of T cells by antigen-presenting cells while circumventing immune checkpoints for enhancing or amplifying a cytotoxic effector cell response. Bulk genomics cannot fully capture the potential of large-scale sequencing data for target identification. This can be carried out effectively using scRNA technology [29]. Since heterogenous cell types are identified by their diverse gene expression patterns, including genomic instability, genetic alterations, and epigenetic modifications, this technique has resulted in the identification of new cellular subtypes whose roles in modulating both the TME and effector immune responses will become clearer in the near future [30]. T-cell receptor sequences hold the key to an effective immune response [31]. In the case of immune responses, a study by Schattgen et al. [32] identified a link between the length of the CDR3 region and the T cell differentiation status. Using the same scRNA technology, it was shown that the bulk of the response against Pneumocystis carinii, which causes PCP, was mounted by CD4+ T lymphocytes [33]. Such crucial links may also be found in the TME and aid in identifying newer targets for eliminating tumour cells.
In addition to target identification, scRNA technology may be useful in evaluating the mechanism of tumour cell response in the face of IMT agents when used alone or as adjuvants to chemotherapy [34]. At a single cell resolution, by detecting the transcriptional activity of immune checkpoint or multidrug resistance genes, the effect of these agents on specific cellular groups can be studied, making them invaluable tools in personalised medicine [9, 12]. Some of the novel applications of scRNA sequencing in immunotherapy are in studying the involvement of the immune system in cancer progression, the impact of drugs on antitumor immunity, and patient treatment outcome and effect [33], [34], [35]. scRNA-seq technology has been applied in the field of drug discovery and development to understand disease subtyping based on cell composition and altered cell states and will help us identify novel cellular and molecular targets for preclinical models [36]. scRNA-seq, along with the functional genomic CRISPR (clustered regulatory interspaced short palindromic repeat) screens, has enabled the exploration of gene function and genetic regulatory networks. This emerging technology for high-throughput investigations facilitates target prioritisation of highly active CRISPR libraries for single-cell screens [37]. Sc-CRISPR screens can assess specific CRISPR-driven gene knockdowns or editing and the resulting perturbed cellular phenotypes in a scalable and comprehensive manner [38].
Current challenges in using scRNA sequencing
scRNA technology is currently available as full-length transcript sequencing or 3′/5′ end sequencing alone. The full-length technologies that are currently available include SmartSeq2, MATQ-Seq, and SUPeR-Seq [39]. For the 3′ end sequencing, Drop Seq, Seq Well, chromium, and DroNc Seq are available. STRT Seq can perform 5′-end sequencing alone. However, currently, scRNA technology suffers from technical and analytical challenges [40, 41]. The technique requires a large amount of starting material and is plagued by low capture and high drop, along with abundant technical noise-related issues. Additionally, it is also limited by biological variations between each cell arising due to the stochastic nature of transcription [40, 41]. The overall quality of the scRNA sequencing data is dependent on the read mapping ratio. No difference exists between scRNA and bulk RNA sequencing data in read alignment. Most of the mapping, expression, and other data analysis tools that have become more streamlined for use across different platforms of bulk RNA sequencing can also be used for scRNA-seq data analysis [41]. However, data bias, such as coverage of the transcripts, capture effectiveness, and sequencing noise resulting from dead or damaged cells, plagues the analysis of scRNA data [42]. Due to such cell quality-related issues, there is the possibility of data misinterpretation. Hence, crucial QC analysis is required to filter this data. Determining the expression levels of rare or abundant events by scRNA-seq still remains a challenge. To some extent, this could be overcome by using the selective enrichment technique, which was developed based on CRISPR technology [43]. Hence, gold-standard technical and computational methods are needed to allow the reproducibility of clinical trials in onco-immunology between centres. Compared to bulk RNA-seq, the cost of performing scRNA-seq is less, but it may still need to be scaled down for use in routine clinical care [44].
Conclusions
Great progress in the past decade has been witnessed in scRNA-seq technology development and innovation. Advances in bioinformatics approaches and artificial intelligence to perform data analysis will facilitate biological and clinical researchers’ understanding of dynamics between distinct cell clusters within tumours, allowing immunotherapy to succeed in treating a variety of malignancies. Inevitably, such strategies may prevent tumour recurrence and increase patient survival.
Acknowledgments
Ahmad Kodous wishes to acknowledge the intellectual and material contributions of DBT and UNESCO-TWAS by awarding him a postdoctoral fellowship at the Molecular Oncology Department, Cancer Institute WIA, Adyar, Chennai, India.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: All authors, Ahmad S. Kodous, Meenakumari Balaiah and Priya Ramanathan, equally contributed to the plan, design, writing, and reviewing of the manuscript.
-
Competing interests: Authors state no conflict of interest.
-
Research funding: None declared.
-
Data availability: Not applicable.
References
1. Xiao, L, Wei, F, Liang, F, Li, Q, Deng, H, Tan, S, et al.. TSC22D2 identified as a candidate susceptibility gene of multi-cancer pedigree using genome-wide linkage analysis and whole-exome sequencing. Carcinogenesis 2019;40:819–27, https://doi.org/10.1093/carcin/bgz095.Search in Google Scholar PubMed
2. Roque, N, Matias, D, Balça-Silva, J, Ferrer, VP, Pessoa, LS, de Sampaio e Spohr, TCL. The interface of cancer, their microenvironment and nanotechnology. Oncologie 2022;24:371–411, https://doi.org/10.32604/oncologie.2022.024035.Search in Google Scholar
3. Álvarez-Arenas, A, Podolski-Renic, A, Belmonte-Beitia, J, Pesic, M, Calvo, GF. Interplay of Darwinian selection, Lamarckian induction and microvesicle transfer on drug resistance in cancer. Sci Rep 2019;9:9332, https://doi.org/10.1038/s41598-019-45863-z.Search in Google Scholar PubMed PubMed Central
4. Cui, X, Peng, M, Chen, H. The emerging role of lymphocyte activation gene 3 in the cancer immunotherapy. Oncologie 2022;24:666–78, https://doi.org/10.32604/oncologie.2022.023641.Search in Google Scholar
5. Ren, D, Hua, Y, Yu, B, Ye, X, He, Z, Li, C, et al.. Predictive biomarkers and mechanisms underlying resistance to PD1/PD-L1 blockade cancer immunotherapy. Mol Cancer 2020;19:1–9, https://doi.org/10.1186/s12943-020-1144-6.Search in Google Scholar PubMed PubMed Central
6. Cao, Y, Qiu, Y, Tu, G, Yang, C. Single-cell RNA sequencing in immunology. Curr Genomics 2020;21:564–75, https://doi.org/10.2174/1389202921999201020203249.Search in Google Scholar PubMed PubMed Central
7. Kuipers, J, Jahn, K, Beerenwinkel, N. Advances in understanding tumour evolution through single-cell sequencing. Biochim Biophys Acta – Rev Cancer 2017;1867:127–38, https://doi.org/10.1016/j.bbcan.2017.02.001.Search in Google Scholar PubMed PubMed Central
8. Wang, M, Gan, J, Han, C, Guo, Y, Chen, K, Shi, YZ, et al.. Imputation methods for scRNA sequencing data. Appl Sci 2022;12:10684, https://doi.org/10.3390/app122010684.Search in Google Scholar
9. Li, L, Xiong, F, Wang, Y, Zhang, S, Gong, Z, Li, X, et al.. What are the applications of single-cell RNA sequencing in cancer research: a systematic review. J Exp Clin Cancer Res 2021;40:1–2, https://doi.org/10.1186/s13046-021-01955-1.Search in Google Scholar PubMed PubMed Central
10. Ahmed, R, Zaman, T, Chowdhury, F, Mraiche, F, Tariq, M, Ahmad, IS, et al.. Single-cell RNA sequencing with spatial transcriptomics of cancer tissues. Int J Mol Sci 2022;23:3042, https://doi.org/10.3390/ijms23063042.Search in Google Scholar PubMed PubMed Central
11. Hong, M, Tao, S, Zhang, L, Diao, LT, Huang, X, Huang, S, et al.. RNA sequencing: new technologies and applications in cancer research. J Hematol Oncol 2020;13:1–6.10.1186/s13045-020-01005-xSearch in Google Scholar PubMed PubMed Central
12. Zhu, S, Qing, T, Zheng, Y, Jin, L, Shi, L. Advances in single-cell RNA sequencing and its applications in cancer research. Oncotarget 2017;8:53763–79, https://doi.org/10.18632/oncotarget.17893.Search in Google Scholar PubMed PubMed Central
13. Hwang, B, Lee, JH, Bang, D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med 2018;50:1–4, https://doi.org/10.1038/s12276-018-0071-8.Search in Google Scholar PubMed PubMed Central
14. Hu, P, Zhang, W, Xin, H, Deng, G. Single cell isolation and analysis. Front Cell Dev Biol 2016;4:116, https://doi.org/10.3389/fcell.2016.00116.Search in Google Scholar PubMed PubMed Central
15. Naeem, A, James, N, Tanvir, M, Marriam, M, Nathaniel, S. Fluorescence activated cell sorting (FACS): an advanced cell sorting technique. PSM Biol Res 2017;2:83–8.Search in Google Scholar
16. Pacheco, A, Blanco, A, Bronet, F, Cruz, M, García-Fernández, J, García-Velasco, JA. Magnetic-activated cell sorting (MACS): a useful sperm-selection technique in cases of high levels of sperm DNA fragmentation. J Clin Med 2020;9:3976, https://doi.org/10.3390/jcm9123976.Search in Google Scholar PubMed PubMed Central
17. Seminati, D, Casati, G, Pagni, F, Fraggetta, F. Laser capture microdissection in lung cancer: a narrative review. J Xiangya Med 2022:1–8, https://doi.org/10.21037/jxym-21-55.Search in Google Scholar
18. Zhang, Q, Gong, H, Ma, J, Li, J, Leng, L. Laser capture microdissection (LCM) of human skin sample for spatial proteomics research. Bio-protocol 2023;13:e4623, https://doi.org/10.21769/bioprotoc.4623.Search in Google Scholar
19. Lohani, V, Akhiya, AR, Kundu, S, Akhter, MQ, Bag, S. Single-cell proteomics with spatial attributes: tools and techniques. ACS Omega 2023;8:17499–510, https://doi.org/10.1021/acsomega.3c00795.Search in Google Scholar PubMed PubMed Central
20. Raj, MK, Chakraborty, S. PDMS microfluidics: a mini review. J Appl Polym Sci 2020;137:48958.10.1002/app.48958Search in Google Scholar
21. Zia, S, Roda, B, Maggio, A, Marrazzo, P, Pizzuti, V, Alviano, F, et al.. Celector®: an innovative technology for quality control of living cells. Appl Sci 2022;12:9967, https://doi.org/10.3390/app12199967.Search in Google Scholar
22. Anaparthy, N, Ho, YJ, Martelotto, L, Hammell, M, Hicks, J. Single-cell applications of next-generation sequencing. Cold Spring Harb Perspect Med 2019;9:a026898, https://doi.org/10.1101/cshperspect.a026898.Search in Google Scholar PubMed PubMed Central
23. Rich-Griffin, C, Stechemesser, A, Finch, J, Lucas, E, Ott, S, Schäfer, P. Single-cell transcriptomics: a high-resolution avenue for plant functional genomics. Trends Plant Sci 2020;25:186–97, https://doi.org/10.1016/j.tplants.2019.10.008.Search in Google Scholar PubMed
24. Clark, IC, Fontanez, KM, Meltzer, RH, Xue, Y, Hayford, C, May-Zhang, A, et al.. Microfluidics-free single-cell genomics with templated emulsification. Nat Biotechnol 2023;41:1–10. https://doi.org/10.1038/s41587-023-01685-z.Search in Google Scholar PubMed PubMed Central
25. Heumos, L, Schaar, AC, Lance, C, Litinetskaya, A, Drost, F, Zappia, L, et al.. Best practices for single-cell analysis across modalities. Nat Rev Genet 2023;24:1–23, https://doi.org/10.1038/s41576-023-00586-w.Search in Google Scholar PubMed PubMed Central
26. Kleino, I, Frolovaitė, P, Suomi, T, Elo, LL. Computational solutions for spatial transcriptomics. Comput Struct Biotechnol J 2022;20:4870–84, https://doi.org/10.1016/j.csbj.2022.08.043.Search in Google Scholar PubMed PubMed Central
27. Feng, H, Lin, L, Chen, J. scDIOR: single cell RNA-seq data IO software. BMC Bioinform 2022;23:1–9, https://doi.org/10.1186/s12859-021-04528-3.Search in Google Scholar PubMed PubMed Central
28. Battenberg, K, Kelly, ST, Ras, RA, Hetherington, NA, Hayashi, M, Minoda, A. A flexible cross-platform single-cell data processing pipeline. Nat Commun 2022;13:6847, https://doi.org/10.1038/s41467-022-34681-z.Search in Google Scholar PubMed PubMed Central
29. Raskov, H, Orhan, A, Christensen, JP, Gögenur, I. Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer 2021;124:359–67, https://doi.org/10.1038/s41416-020-01048-4.Search in Google Scholar PubMed PubMed Central
30. Park, JW, Han, JW. Targeting epigenetics for cancer therapy. Arch Pharm Res 2019;42:159–70, https://doi.org/10.1007/s12272-019-01126-z.Search in Google Scholar PubMed PubMed Central
31. Wang, D, Chen, H, Hu, Y. Polarized autologous macrophages (PAM) can Be a tumor vaccine. Oncologie 2022;24:441–9, https://doi.org/10.32604/oncologie.2022.024898.Search in Google Scholar
32. Schattgen, SA, Guion, K, Crawford, JC, Souquette, A, Barrio, AM, Stubbington, MJT, et al.. Integrating T cell receptor sequences and transcriptional profiles by clonotype neighbor graph analysis (CoNGA). Nat Biotechnol 2022;40:54–63, https://doi.org/10.1038/s41587-021-00989-2.Search in Google Scholar PubMed PubMed Central
33. Xie, J, Chen, L, Sun, Q, Li, H, Wei, W, Wu, D, et al.. An immune subtype-related prognostic signature of hepatocellular carcinoma based on single-cell sequencing analysis. Aging 2022;14:3276, https://doi.org/10.18632/aging.204012.Search in Google Scholar PubMed PubMed Central
34. Sun, G, Li, Z, Rong, D, Zhang, H, Shi, X, Yang, W, et al.. Single-cell RNA sequencing in cancer: applications, advances, and emerging challenges. Mol Ther – Oncolytics 2021;21:183–206, https://doi.org/10.1016/j.omto.2021.04.001.Search in Google Scholar PubMed PubMed Central
35. Zhang, Y, Zhang, Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol 2020;17:807–21, https://doi.org/10.1038/s41423-020-0488-6.Search in Google Scholar PubMed PubMed Central
36. Kalimuthu, S, Jeong, JH, Oh, JM, Ahn, BC. Drug Discovery by molecular imaging and monitoring therapy response in lymphoma. Int J Mol Sci 2017;18:1639, https://doi.org/10.3390/ijms18081639.Search in Google Scholar PubMed PubMed Central
37. Castells-Roca, L, Tejero, E, Rodríguez-Santiago, B, Surrallés, J. Crispr screens in synthetic lethality and combinatorial therapies for cancer. Cancers 2021;13:1591, https://doi.org/10.3390/cancers13071591.Search in Google Scholar PubMed PubMed Central
38. Qiu, Z, Wang, Y, Zhang, Z, Qin, R, Peng, Y, Tang, W, et al.. Roles of intercellular cell adhesion molecule-1 (ICAM-1) in colorectal cancer: expression, functions, prognosis, tumorigenesis, polymorphisms and therapeutic implications. Front Oncol 2022;12:1052672, https://doi.org/10.3389/fonc.2022.1052672.Search in Google Scholar PubMed PubMed Central
39. Hahaut, V, Picelli, S. Full-length single-cell RNA-sequencing with FLASH-seq. In: Single cell transcriptomics: methods and protocols. New York, NY: Springer US; 2022:123–64 pp.10.1007/978-1-0716-2756-3_5Search in Google Scholar PubMed
40. Ding, J, Adiconis, X, Simmons, SK, Kowalczyk, MS, Hession, CC, Marjanovic, ND, et al.. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat Biotechnol 2020;38:737–46, https://doi.org/10.1038/s41587-020-0465-8.Search in Google Scholar PubMed PubMed Central
41. Chen, G, Ning, B, Shi, T. Single-cell RNA-seq technologies and related computational data analysis. Front Genet 2019;10:317, https://doi.org/10.3389/fgene.2019.00317.Search in Google Scholar PubMed PubMed Central
42. Lähnemann, D, Köster, J, Szczurek, E, McCarthy, DJ, Hicks, SC, Robinson, MD, et al.. Eleven grand challenges in single-cell data science. Genome Biol 2020;21:1–35, https://doi.org/10.1186/s13059-020-1926-6.Search in Google Scholar PubMed PubMed Central
43. Meza-Sosa, KF, Miao, R, Navarro, F, Zhang, Z, Zhang, Y, Hu, JJ, et al.. SPARCLE, a p53-induced lncRNA, controls apoptosis after genotoxic stress by promoting PARP-1 cleavage. Mol Cell 2022;82:785–802, https://doi.org/10.1016/j.molcel.2022.01.001.Search in Google Scholar PubMed PubMed Central
44. Kuksin, M, Morel, D, Aglave, M, Danlos, FX, Marabelle, A, Zinovyev, A, et al.. Applications of single-cell and bulk RNA sequencing in onco-immunology. Eur J Cancer 2021;149:193–210, https://doi.org/10.1016/j.ejca.2021.03.005.Search in Google Scholar PubMed
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism
Articles in the same Issue
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism